Back to Journals » Drug Design, Development and Therapy » Volume 19
Specnuezhenide Alleviates Senile Osteoporosis by Activating TGR5/FXR Signaling in Bone Marrow Mesenchymal Stem Cells and RANKL-Induced Osteoclasts
Authors Deng X, Lin B, Xiao W, Wang F, Xu P, Wang N
Received 30 August 2024
Accepted for publication 27 February 2025
Published 6 March 2025 Volume 2025:19 Pages 1595—1608
DOI https://doi.org/10.2147/DDDT.S493711
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Professor Anastasios Lymperopoulos
Xuehui Deng,1,2,* Bingfeng Lin,1,* Wenlong Xiao,3 Fang Wang,2 Pingcui Xu,1 Nani Wang1– 3
1Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, People’s Republic of China; 2School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China; 3School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Nani Wang, Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, 132 Tianmushan Road, Hangzhou, Zhejiang, 310007, People’s Republic of China, Tel/Fax +86-571-88849089, Email [email protected]
Background: Specnuezhenide (SPN) is an iridoid glycoside isolated from Fructus Ligustri Lucidi, an herb prescribed for the treatment of senile osteoporosis. However, the direct role of SPN on bone metabolism remains unclear. In this study, the effects of SPN on
Methods: Micro-computed tomography was used to observe the bone microstructure. Osteogenesis was examined using Western blotting and alkaline phosphatase staining. Osteoclastogenesis was examined using Western blotting and F-actin ring staining. Senescence-associated β-galactosidase was used to detect cell senescence. In addition, the expression of Takeda G protein-coupled receptor 5 (TGR5)/farnesoid X receptor (FXR) signaling pathway-related genes and proteins was determined through quantitative real-time polymerase chain reaction and immunofluorescence.
Results: Oral administration of SPN improved the bone microstructure in
Conclusion: SPN alleviated senile osteoporosis and cell senescence by activating the TGR5/FXR pathway.
Keywords: specnuezhenide, Takeda G protein-coupled receptor 5, farnesoid X receptor, osteoporosis
Introduction
Senile osteoporosis is characterized by deterioration of the bone microstructure and reduced bone strength, resulting in a high fracture risk.1 With the dramatic increase in the global population of older adults, this disease has emerged as a public health challenge worldwide.2,3 Bone loss can directly lead to bone fragility, and the cause of bone loss during aging is closely related to the aging of bone marrow mesenchymal stem cells (BMSCs).4,5 BMSCs have strong self-renewal ability and multidirectional differentiation potential, and can differentiate into a variety of cells, including osteoblasts.6,7 In addition to the osteogenic ability of osteoblasts, the normal growth of bone is also dependent on the bone resorption of osteoclasts.8 Senescent-cell conditioned media have been shown to impair enhanced osteoclast-progenitor survival, leading to increased osteoclastogenesis.9 The expression of the bile acid receptors Takeda G protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR) is downregulated in the aging bone. TGR5 knockdown has been shown to induce osteoclast differentiation to decrease bone mass in aged mice,10 while activation of FXR promotes the differentiation of BMSCs into osteoblasts.11 FXR agonists have also been shown to suppress osteoclast differentiation from bone marrow macrophages.12 A growing body of evidence has shown that TGR5 and FXR are important regulators in the development of senile osteoporosis. Thus, the TGR5/FXR signaling pathway is a promising target for mitigating senile osteoporosis.
Specnuezhenide (SPN) is an iridoid glycoside isolated from Fructus Ligustri Lucidi,13 which is clinically used in the treatment of senile osteoporosis.14,15 Administration of the extract of Fructus Ligustri Lucidi has been shown to restore bone mass in
Materials and Methods
Mice and Treatment
Male ICR mice (age, 8 weeks; weight, 20–22 g) were obtained from Hangzhou Medical College (Hangzhou, Zhejiang, China). All animal experiments were conducted in accordance with the NIH Guide for the Care and Use of Laboratory Animals (NIH Publication No. 80–23; revised 1978) and the Guidelines for Animal Experiments of the Zhejiang Academy of Traditional Chinese Medicine (Approval No. 2021–001). The protocol was approved by the Institutional Animal Ethics Committee. SPN (#102728; purity ≥ 98%; Yongjian Pharmaceutical, Jiangsu, China; Figure 1A) was dissolved in sterile saline. The mice were randomly divided into the following groups: normal control group (CON: daily gastric irrigation [i.g.] of sterile saline + daily subcutaneous injection [s.q.] of sterile saline, n = 10), model group (MOD: daily i.g. of sterile saline + daily s.q. of 150 mg/kg/d
Micro-Computed Tomography Bone Analysis
Femurs were removed and kept in 4% formalin. Fixed femurs were scanned using a micro-computed tomography (micro-CT) scanner (Skyscan 1172; Bruker, Belgium). Values for bone mineral density (BMD), bone volume/total volume (BV/TV), trabecular thickness (Tb.Th), and trabecular number (Tb.N) were obtained.
Hematoxylin-Eosin Staining
Femurs were embedded in paraffin. Hematoxylin-eosin (HE) staining was performed using a standard protocol reported previously.13 The sections were stained with an HE assay kit (#G1076; Servicebio, Hubei, China).
Dynamic Histomorphometric Assessments of Bone Formation
The dynamic histomorphometry parameters measured included mineral apposition rate (MAR).21 The mice were injected with calcein (35 mg/kg, intraperitoneal injection [i.p.]; Sangon Biotech, Shanghai, China) and tetracycline (20 mg/kg, i.p.; Aladdin, Shanghai, China) dissolved in sterile saline at 10 and 2 days before harvesting the femurs. The femurs were embedded in methyl methacrylate (#M813511; Macklin, Shanghai, China) to form blocks, and sectioned into 10-μm slices. The sections were viewed under a fluorescent Pannoramic MIDI system (3DHISTECH).
Western Blotting Analysis
The tibia was homogenized and extracted in RIPA lysis buffer (Beyotime, Shanghai, China). The protein content was measured using a BCA kit (Keygentec, Nanjing, China). The primary antibodies included anti-FXR (#DF12402; Affinity, Jiangsu, China), anti-TGR5 (#DF14087; Affinity), anti-small heterodimer partner (SHP, #AF6244; Affinity), anti-alkaline phosphatase (ALP, #BM4284; Boster, Hubei, China), anti-runt-related transcription factor 2 (Runx2, #DF0171; Boster), anti-bone morphogenetic protein 2 (BMP2, #AF5163; Affinity), anti-nuclear factor-κB (RANK, #DF12532; Affinity), and anti-nuclear factor of activated T-cells (NFATc1, #D0522; Santa Cruz Biotechnology). The membrane was incubated with goat anti-rabbit IgG. Protein band densities were analyzed using ImageJ software and normalized to the GAPDH level.
Immunohistochemistry
Femurs were collected, fixed with 4% formalin for 48 h, decalcified in 10% ethylenediaminetetraacetic acid (EDTA), paraffin-embedded, and used for tartrate-resistant acid phosphatase (TRAP) and Masson’s trichrome staining.22,23 Next, 5-μm sections were incubated with primary antibodies against osteocalcin (OCN, #DF12303; Boster), TGR5, FXR, P16 (#AF0228, Affinity), P21 (#DF6423, Affinity), P53 (#AF0879, Affinity), and CD146 (#17564-1-AP, Proteintech). The images were analyzed by Image-Pro Plus 6.0 software (Media Cybernetics, Silver Spring, USA).
Cell Culture and Treatment
BMSCs and RAW264.7 cells were purchased from the National Collection of Authenticated Cell Cultures (Shanghai, China). Cells were cultured in Dulbecco’s modified essential medium supplemented with 10% fetal bovine serum and 1% antibiotics, and cultured at 37°C in a humidified atmosphere containing 5% CO2 until confluence. To generate osteoclasts, RAW264.7 cells (3 × 103 cells/well) were cultured in the presence of 50 ng/mL nuclear factor-κB ligand (RANKL) and various additives. BMSCs and OC were respectively divided into the following groups: blank control group,
SA-β-gal Staining
SA-β-gal staining was performed in accordance with the manufacturer’s instructions (#C0602, Beyotime). Briefly, cells were fixed in 4% paraformaldehyde for 15 min and incubated with SA-β-gal staining solution at 37°C for 12 h. The cells were washed with phosphate-buffered saline (PBS) and photographed under a microscope (TI-DH; Nikon, Tokyo, Japan). The percentage of SA-β-gal-positive cells was expressed as the ratio of blue-stained cells to the total number of cells.
ALP Staining
After the BMSCs reached 80% confluence, the medium was replaced with an osteogenic induction medium (100 nM dexamethasone, 10 mm β-glycerophosphate, and 50 μM ascorbic acid).
F-Actin Ring Staining
After 5 d of OC induction, the cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 in PBS for 5 min, and then stained with fluorescein isothiocyanate (FITC)-phalloidin solution (#C1033, Beyotime) in darkness for 1 h. After washing with PBS, the cells were incubated with a 4′,6-diamidino-2-phenylindole (DAPI) solution for 10 min to stain the nuclei. The images of F-actin rings were visualized under a confocal microscope (LSM800; Zeiss, Oberkochen, Germany).
RNA Sequencing
RNA was isolated using TRIzol reagent. Sequencing libraries were generated using the ALFA-SEQ Directional RNaLib Prep Kit (mChip, Guangzhou, China). The mRNA was purified using poly-T oligo-attached magnetic beads. First-strand cDNA was prepared using random hexamer primers and M-MuLV reverse transcriptase (RNase H). Second-strand cDNA was synthesized using DNA polymerase I and RNase H. Library quality was assessed on a Qsep400 high-throughput nucleic acid protein analysis system (Houze Biotechnology Co., Ltd., Hangzhou, China). Genes were considered to be differentially expressed when the fold changes were ≥2 and the corrected P value was <0.05.
Immunofluorescence Staining
Cells were fixed with 4% paraformaldehyde for 15 min and permeabilized with 0.1% Triton X-100 (this step was omitted for TGR5). Cells were incubated with anti-TGR5 (dilution 1:500) or anti-FXR (dilution 1:500) antibodies at 4°C for 12 h, and then incubated with goat pAb to Rb lgG (Alexa Fluor® 488, #ab150077, Abcam) at room temperature for 1 h. The slides were mounted with DAPI and photographed under a confocal microscope.
RNA Isolation and Real-Time Reverse Transcription Polymerase Chain Reaction
Total RNA was extracted from tibias using TRIzol (Invitrogen, Carlsbad, CA, US). The RNA sample was reverse-transcribed using a TOBOBlue qRT Premix with a gDNA Eraser 2.0 kit (#RTQ202, Toroivd Technology, Shanghai, China). The mRNA expression levels of SHP, TGR5, and FXR were measured in a 7500 quantitative real-time reverse transcription polymerase chain reaction system (RT-PCR; Applied Biosystems, Waltham, MA, US) using SYBR Green reagents (#QPS201; Toyobo, Osaka, Japan). The primer sequences are listed in Table S1. Relative mRNA expression was quantified by comparing cycle threshold values. GAPDH was used as a housekeeping gene, and data were presented as the fold change relative to the control.
Statistical Analysis
Statistical analysis was performed using GraphPad Prism 9.0 (GraphPad, CA, US). Data were presented as the mean ± standard deviation. Statistical comparisons were performed using one-way or two-way analysis of variance followed by Dunnett corrections to compare multiple groups. P < 0.05 was considered statistically significant.
Results
SPN Promotes Osteoporosis in d-gal-Induced Mice
To determine the effect of SPN on bone microstructure, micro-CT analysis was performed on mouse femurs. The SPN-L and SPN-H groups showed higher BMD (Figure 1C, P < 0.01), BV/TV (P < 0.01), Tb.N (P < 0.01), and Tb.Th (P < 0.01) in comparison with the corresponding values in the MOD group. HE staining showed that the cancellous bone in the femur in the
SPN Increases Bone Formation in d-gal-Induced Mice
OCN is specifically secreted by osteoblasts and is considered an essential biomarker of bone formation.26 The expression of OCN was downregulated in the MOD group in comparison with that in the CON group (Figure 2A, P < 0.01), while SPN restored OCN expression (P < 0.01). SPN also promoted the expression of ALP (Figure 2B, P < 0.05 for SPN-L and P < 0.01 for SPN-H), BMP2 (P < 0.01 for SPN-H), and Runx2 (P < 0.01) in
SPN Decreases Bone Resorption in d-gal-Induced Mice
TRAP staining was performed to examine whether SPN alleviated
SPN Attenuates Bone Senescence in d-gal-Induced Mice
Cellular senescence is a master issue in the development of age-related osteoporosis, so we measured the effect of SPN on senescence markers in mouse femurs. The expression levels of the senescence-related proteins P16, P21, and P53 were significantly higher in model mice (Figure 4A and B, P < 0.01) in comparison with those in the control group. SPN downregulated the expression levels of these proteins in comparison with those in the model group in a dose-dependent manner (P < 0.01).
SPN Attenuates Cell Senescence Through the TGR5/FXR Pathway
To study the effects of SPN on BMSCs and RAW264.7 cells, we detected the proliferation rates of the two kinds of cells under different concentrations of SPN, and found that the proliferation rates of the two kinds of cells under different concentrations of SPN did not differ from those in the control group (Figure S2A and B), suggesting that SPN was not toxic to cells. We also detected SA-β-gal-positive cells to evaluate the anti-senescence effects of SPN. As expected,
SPN Promoted Osteogenic Differentiation of BMSCs and Blocked Osteoclast Differentiation of RAW264.7 Cells Through the TGR5/FXR Pathway
BMSCs are identified by detection of CD146, a mesenchymal stem cell marker (Figure S3).27 ALP is an early-stage osteogenic-differentiation marker.28 The osteogenic induction medium induced osteogenic differentiation of BMSCs along with upregulation of ALP expression. SPN also increased the expression of ALP (Figure 6A). To confirm whether activation of TGR5 and FXR can induce ALP generation in BMSCs, we detected the ALP level in BMSCs after stimulation with the TGR5 antagonist SBI-115 or FXR antagonist DY268. SBI-115 and DY268 suppressed ALP expression. As a result, the osteogenic-differentiation effect of SPN was blocked in the presence of the SBI-115 or DY268.
![]() |
Figure 6 SPN promoted of ALP expression and inhibited of F-actin formation. (A) ALP staining. (B) F-actin staining. |
Formation of F-actin-rich adhesive structures by osteoclasts is an essential step in bone resorption.29 RANKL stimulation increased well-defined F-actin sealing rings with greater ring heights. The F-actin rings in the MOD group showed a typically huge ring structure (Figure 6B), those in the SPN group were obviously smaller. RANKL-induced RAW264.7 cells were also abrogated by the TGR5 antagonist SBI-115 or FXR antagonist DY268. The TRAP results showed that in comparison with the control group, the number and area of OCs in the model group were higher, while SPN administration alleviated this phenomenon (Figure S4). These results indicate that SPN can inhibit OC differentiation.
SPN Upregulates TGR5 and FXR Expression in d-gal-Induced BMSCs and OCs
Next, we performed BMSC RNA-sequence analysis to explore the molecular mechanisms underlying the effects of SPN. In comparison with the expression levels in the model group, FXR predominated the upregulated genes (Figure 7A). Gene set enrichment analysis (GSEA) also revealed that SPN could regulate bile acid biosynthetic process-related genes (Figure 7B and C). To confirm whether SPN functions through TGR5-FXR activation, we evaluated TGR5 and FXR expression in vitro (Figure 7D and E).
SPN Activates TGR5 and FXR in d-gal-Induced Mice
We next examined the effects of SPN on TGR5 and FXR expression in bone tissues. The protein levels of TGR5 (Figure 8A, P < 0.01) and FXR (P < 0.01) were downregulated in the MOD group in comparison with the CON group, while SPN increased TGR5 and FXR expression (P < 0.01). Western blot results showed that SPN significantly upregulated the protein expression levels of TGR5 (Figure 8B, P < 0.01), FXR (P < 0.01), and its downstream SHP (P < 0.01) in mouse tibias in comparison with those in the MOD group.
SPN treatment also upregulated the mRNA expression of TGR5 (Figure 8C, P < 0.01), FXR (P < 0.01), and SHP (P < 0.01) in
Discussion
A variety of active ingredients have been identified in Fructus Ligustri Lucidi. Previous investigations by our group and other researchers have shown that the aqueous extract and some compounds of Fructus Ligustri Lucidi are beneficial for bone metabolism.30 However, the direct osteoprotective effects of SPN, the major active component of Fructus Ligustri Lucidi, in aging models remain to be elucidated. The present study is the first to demonstrate that administration of SPN increased the bone mass of
TGR5 and FXR are bile acid receptors that serve as essential regulators in the development of senile osteoporosis.34,35 TGR5 deletion has been shown to markedly decrease bone mass in aged and ovariectomized mice.6 Upregulation of TGR5 can promote osteoblast mineralization and increase the expression of osteoblast differentiation marker genes.31 TGR5 regulates osteoclast differentiation to reduce bone loss.36 FXR has also been shown to suppress osteoclast differentiation and promote osteoblast differentiation.11 FXR typically heterodimerizes with the retinoid X receptor in FXR response elements associated with the promoters of the target gene Runx2, which is a bone-specific transcription factor. Deletion of FXR in vivo leads to significant bone loss.37 In this study, we observed that SPN increased the levels of TGR5 and FXR and the bone formation biomarkers Runx2, OCN, BMP2, and ALP in
Conclusions
In summary, this study is the first to demonstrate that SPN alleviates senile osteoporosis. This effect was associated with activation of the TGR5/FXR pathway, stimulation of osteoblastic differentiation of BMSCs, inhibition of osteoclastic differentiation of RANKL-induced RAW264.7 cells, and attenuation of cell senescence. Future studies should further evaluate the toxicity of this compound and investigate its anti-osteoporosis effects in females. These findings provide a theoretical basis for the use of SPN as a functional supplement in preventing and treating osteoporosis.
Abbreviations
Alkaline phosphatase (ALP), bone mesenchymal stem cells (BMSCs), bone mineral density (BMD), bone morphogenetic protein 2 (BMP2), bone volume/total volume (BV/TV), D-galactose (D-gal), ethylenediamine tetraacetic acid (EDTA), farnesoid X receptor (FXR), gene set enrichment analysis (GSEA), gastric irrigation (ig), immunohistochemistry (IHC), micro-computed tomography (micro-CT), osteoclast (OC), osteocalcin (OCN), nuclear factor-κB ligand (RANKL), nuclear factor-κB (RANK), nuclear factor of activated T-cells (NFATc1), real-time reverse transcription polymerase chain reaction (RT-PCR), runt-related transcription factor 2 (Runx2), subcutaneous injection (sq), senescence-associated β-galactosidase (SA-β-gal), small heterodimer partner (SHP), specnuezhenide (SPN), Takeda G protein receptor 5 (TGR5), trabecular number (Tb.N), trabecular thickness (Tb.Th), Tartrate-resistant acid phosphatase (TRAP).
Funding
This work was supported by National Natural Science Foundation of China (82374012), Zhejiang Provincial Natural Science Foundation of China (LHDMZ25H280004), Zhejiang Provincial Medicine Foundation (2022ZX002, 2021ZYY28, GZY-ZJ-KJ-23006, 2024ZR001), and Zhejiang Province Chai Xiujuan Famous old TCM expert inheritance studio construction project.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Qadir A, Liang S, Wu Z, et al. Senile osteoporosis: the involvement of differentiation and senescence of bone marrow stromal cells. Int J mol Sci. 2020;21:349. doi:10.3390/ijms21010349
2. Guo Y, Jia X, Cui Y, et al. Sirt3-mediated mitophagy regulates AGEs-induced BMSCs senescence and senile osteoporosis. Redox Biol. 2021;41:101915. doi:10.1016/j.redox.2021.101915
3. Jiang YY, Li JD, Xue X, et al. Engineered extracellular vesicles for bone therapy. Nano Today. 2022;44:101487. doi:10.1016/j.nantod.2022.101487
4. Hu M, Xing L, Zhang L, et al. NAP1L2 drives mesenchymal stem cell senescence and suppresses osteogenic differentiation. Aging Cell. 2022;21:e13551. doi:10.1111/acel.13551
5. Guo J, Wang F, Hu Y, et al. Exosome-based bone-targeting drug delivery alleviates impaired osteoblastic bone formation and bone loss in inflammatory bowel diseases. Cell Rep Med. 2023;4:100881. doi:10.1016/j.xcrm.2022.100881
6. Zeng ZL, Xie H. Mesenchymal stem cell-derived extracellular vesicles: a possible therapeutic strategy for orthopaedic diseases: a narrative review. Biomater Transl. 2022;3:175–187. doi:10.12336/biomatertransl.2022.03.002
7. Deng S, Zhu F, Dai K, et al. Harvest of functional mesenchymal stem cells derived from in vivo osteo-organoids. Biomater Transl. 2023;4:270–279. doi:10.12336/biomatertransl.2023.04.006
8. Wang J, Li X, Wang S, et al. Bone-targeted exosomes: strategies and applications. Adv Healthc Mater. 2023;12:e2203361. doi:10.1002/adhm.202203361
9. Farr JN, Xu M, Weivoda MM, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017;23:1072–1079. doi:10.1038/nm.4385
10. Li L, Chen B, Zhu R, et al. Fructus Ligustri Lucidi preserves bone quality through the regulation of gut microbiota diversity, oxidative stress, TMAO and Sirt6 levels in aging mice. Aging. 2019;11:9348–9368. doi:10.18632/aging.102376
11. Boufker HI, Lagneaux L, Fayyad-Kazan H, et al. Role of farnesoid X receptor (FXR) in the process of differentiation of bone marrow stromal cells into osteoblasts. Bone. 2011;49:1219–1231. doi:10.1016/j.bone.2011.08.013
12. Cho SW, An JH, Park H, et al. Positive regulation of osteogenesis by bile acid through FXR. J Bone Miner Res. 2013;28:2109–2121. doi:10.1002/jbmr.1961
13. Wu J, Ke X, Fu W, et al. Inhibition of hypoxia-induced retinal angiogenesis by specnuezhenide, an effective constituent of Ligustrum Lucidum Ait. through suppression of the HIF-1α/VEGF signaling pathway. Molecules. 2016;21:1756. doi:10.3390/molecules21121756
14. Chen B, Wang L, Li L, et al. Fructus Ligustri Lucidi in osteoporosis: a review of its pharmacology, phytochemistry, pharmacokinetics and safety. Molecules. 2017;22:1469. doi:10.3390/molecules22091469
15. Hu D, Huang S, Ding Y, et al. Specnuezhenide reduces carbon tetrachloride-induced liver injury in mice through inhibition of oxidative stress and hepatocyte apoptosis. J Pharm Pharmacol. 2022;74:191–199. doi:10.1093/jpp/rgab164
16. Li Z, Huang J, Wang F, et al. Dual targeting of bile acid receptor-1 (TGR5) and farnesoid X receptor (FXR) prevents estrogen-dependent bone loss in mice. J Bone Miner Res. 2019;34(4):765–776. doi:10.1002/jbmr.3652
17. Ye X, Jiang J, Yang J, et al. Specnuezhenide suppresses diabetes-induced bone loss by inhibiting RANKL-induced osteoclastogenesis. Acta Biochim Biophys Sin. 2022;54:1080–1089. doi:10.3724/abbs.2022094
18. Dell RB, Holleran S, Ramakrishnan R. Sample size determination. ILAR J. 2002;43(4):207–213. doi:10.1093/ilar.43.4.207
19. Ma C, Zhou X, Xu K, et al. Specnuezhenide decreases interleukin-1β-induced inflammation in rat chondrocytes and reduces joint destruction in osteoarthritic rats. Front Pharmacol. 2018;9:700. doi:10.3389/fphar.2018.00700
20. Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7:27–31. doi:10.4103/0976-0105.177703
21. Chin KY, Abdul-Majeed S, Mohamed N, et al. The effects of tocotrienol and lovastatin co-supplementation on bone dynamic histomorphometry and bone morphogenetic protein-2 expression in rats with estrogen deficiency. Nutrients. 2017;9:143. doi:10.3390/nu9020143
22. Ge Q, Yang S, Qian Y, et al. Ambient PM2.5 exposure and bone homeostasis: analysis of UK biobank data and experimental studies in mice and in vitro. Environ Health Perspect. 2023;131:107002. doi:10.1289/EHP11646
23. Hu L, Xie X, Xue H, et al. MiR-1224-5p modulates osteogenesis by coordinating osteoblast/osteoclast differentiation via the Rap1 signaling target ADCY2. Exp Mol Med. 2022;54:961–972. doi:10.1038/s12276-022-00799-9
24. Wang Y, Hu X, Zhang L, et al. Bioinspired extracellular vesicles embedded with black phosphorus for molecular recognition-guided biomineralization. Nat Commun. 2019;27:2829. doi:10.1038/s41467-019-10761-5
25. Xu J, Wang Y, Li J, et al. IL-12p40 impairs mesenchymal stem cell-mediated bone regeneration via CD4+ T cells. Cell Death Differ. 2016;23:1941–1951. doi:10.1038/cdd.2016.72
26. Rifai OA, Chow J, Lacombe J, et al. Proprotein convertase furin regulates osteocalcin and bone endocrine function. J Clin Invest. 2017;127:4104–4117. doi:10.1172/JCI93437
27. Sasao T, Fukuda Y, Yoshida S, et al. Population doubling level-dependent change of secreted glycosaminoglycan in equine bone marrow-derived mesenchymal stem cells. J Equine Sci. 2015;26:73–80. doi:10.1294/jes.26.73
28. Zhang J, Zhang W, Dai J, et al. Overexpression of Dlx2 enhances osteogenic differentiation of BMSCs and MC3T3-E1 cells via direct upregulation of osteocalcin and ALP. Int J Oral Sci. 2019;11:12. doi:10.1038/s41368-019-0046-1
29. Jeong JW, Ji SY, Lee H, et al. Fermented sea tangle (Laminaria japonica Aresch) suppresses RANKL-induced osteoclastogenesis by scavenging ROS in RAW 264.7 cells. Foods. 2019;8:290. doi:10.3390/foods8080290
30. Wang C, Gao H, Cai E, et al. Protective effects of Acanthopanax senticosus - Ligustrum lucidum combination on bone marrow suppression induced by chemotherapy in mice. Biomed Pharmacother. 2019;109:2062–2069. doi:10.1016/j.biopha.2018.11.071
31. Xu P, Lin B, Deng X, et al. VDR activation attenuates osteoblastic ferroptosis and senescence by stimulating the Nrf2/GPX4 pathway in age-related osteoporosis. Free Radic Biol Med. 2022;193:720–735. doi:10.1016/j.freeradbiomed.2022.11.013
32. El-Baz FK, Saleh DO, Abdel jaleel GA, et al. Heamatococcus pluvialis ameliorates bone loss in experimentally-induced osteoporosis in rats via the regulation of OPG/RANKL pathway. Biomed Pharmacother. 2019;116:109017. doi:10.1016/j.biopha.2019.109017
33. Yang Q, Zou Y, Wei X, et al. PTP1B knockdown alleviates BMSCs senescence via activating AMPK-mediated mitophagy and promotes osteogenesis in senile osteoporosis. Biochim Biophys Acta Mol Basis Dis. 2023;1869:166795. doi:10.1016/j.bbadis.2023.166795
34. Wang Q, Wang G, Wang B, et al. Activation of TGR5 promotes osteoblastic cell differentiation and mineralization. Biomed Pharmacother. 2018;108:1797–1803. doi:10.1016/j.biopha.2018.08.093
35. Fujimori K, Iguchi Y, Yamashita Y, et al. Synthesis of novel farnesoid X receptor agonists and validation of their efficacy in activating differentiation of mouse bone marrow-derived mesenchymal stem cells into osteoblasts. Molecules. 2019;24:4155. doi:10.3390/molecules24224155
36. Zhang Y, Wei J, Feng X, et al. Folic acid supplementation prevents high body fat-induced bone loss through TGR5 signaling pathways. Food Funct. 2024;15:4193–4206. doi:10.1039/D4FO00404C
37. Liu M, Jin F, Zhang S, et al. Activation of farnesoid X receptor signaling by geniposidic acid promotes osteogenesis. Phytomedicine. 2022;103:154258. doi:10.1016/j.phymed.2022.154258
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.